+ All Categories
Home > Documents > Serotonin in the Modulation of Neural Plasticity and Networks. Implications for Neurodevelopmental...

Serotonin in the Modulation of Neural Plasticity and Networks. Implications for Neurodevelopmental...

Date post: 09-Mar-2016
Category:
Upload: karla-segoviano
View: 10 times
Download: 0 times
Share this document with a friend
Description:
Serotonin in the Modulation of Neural Plasticity and Networks. Implications for Neurodevelopmental Disorders

of 17

Transcript
  • Neuron

    wl

    cWie

    lonfrnftn

    widely distributed transmitter in the brain (Dahlstrom and Fuxe,

    5-HT is involved in basic morphogenetic activities during brain

    processes, which supports competent social functioning, has

    neural circuits to process and refine information. Thus, identifi-Considerable evidence links dysfunction of 5-HT transmission

    to neurodevelopmental and subsequent psychiatric disorders

    characterized by compromised function of the so-called social

    the brain 5-HT system and then discuss how 5-HT shapes brain

    networks during development and modulates a spectrum of

    essential neuronal functions. We will consider the current under-brain (Blakemore, 2008; Frith and Frith, 2012). The social brain

    is a construct comprising representations of internal somatic

    standing of how 5-HT receptor-mediated molecular mecha-

    nisms contribute to neuronal development, synapse formationdevelopment and in the life-spanning adaptive capacity of the

    brain, including modulation of neural cell proliferation, migration

    and differentiation as well as neurite outgrowth, axonal guid-

    ance, synaptogenesis, and efficiency of transsynaptic signaling

    (for review, Azmitia and Whitaker-Azmitia, 1997; Daubert and

    Condron, 2010; Gaspar et al., 2003).

    cation of genetic variation affecting molecules essential for the

    formation, specification, and function of excitatory and inhibitory

    synapses is expanding research efforts in neurodevelopmental

    disorders characterized by deficits in attention, motivation,

    cognition, and emotion.

    Here, we will first describe selected fundamental features of1964; Steinbusch, 1981). 5-HT signaling pathways integrate

    not only basic physiology, but also essential brain functions,

    including sensory processing, cognitive control, emotion regula-

    tion, autonomic responses, and motor activity in an overarching

    fashion. Vice versa, it is a target of many physiologic regulators,

    including modulators of gene transcription, neurotrophic pep-

    tides, and steroids as well as psychotropic therapeutics, which

    impact the formation and activity of 5-HT subsystems.

    Brain function is ultimately contingent on a specific patterning

    of connections between distinct populations of neurons and the

    establishment of functional neural circuits. The strength of

    synaptic signals is continuously modified by sensory input,

    event-related neural activity and experience, adaptive pro-

    cesses commonly referred to as synaptic plasticity. The molec-

    ular, cellular and circuitry changes associated with synaptic

    plasticity are believed to moderate higher-order brain tasks,

    such as social cognition and emotional learning and memory.

    recently been associated with activity in distinct neural circuits,

    including networks involved in imitation, imitative learning, social

    cognition, and communication skills (Amodio and Frith, 2006;

    Carr et al., 2003). Deficits in 5-HT-moderated synaptic signaling

    resulting in impairments of these network processes fundamen-

    tally impact pathophysiology and long-term outcome of neuro-

    developmental disorders comprising a spectrum of diseases

    ranging from schizophrenic psychoses to autism spectrum and

    attention-deficit/hyperactivity disorders.

    In addition to the role in synapse formation and network

    construction during development, increasing evidence impli-

    cates 5-HT in the regulation of cell adhesion molecules critically

    involved in the plasticity of the developing and adult brain (Dalva

    et al., 2007; Yamagata et al., 2003). These synaptic adhesion

    molecules are principal components of the molecular machinery

    that connects pre- and postsynaptic neurons, facilitates trans-

    mission, controls synaptic plasticity, and empowers intersectingReview

    Serotonin in the Modulationof Neural Plasticity and NetImplications for Neurodeve

    Klaus-Peter Lesch1,2,* and Jonas Waider11Division of Molecular Psychiatry, Laboratory of Translational NeurosPsychosomatics and Psychotherapy, University of Wurzburg, 970802Department of Neuroscience, School of Mental Health and Neurosc*Correspondence: [email protected]://dx.doi.org/10.1016/j.neuron.2012.09.013

    Serotonin (5-HT) shapes brain networks during deveneuronal functions ranging from perception and cogbrain. Deficits in 5-HT-moderated synaptic signalingterm outcome of neurodevelopmental disorders. Ouof circuit configuration influences social cognition ainsight into the molecular and cellular mechanisms odiscuss emerging concepts as to howdefects in synapfindings regarding 5-HTs role in brain development aiological basis of neurodevelopmental disorders.

    IntroductionSerotonin (5-hydroxtryptamine, 5-HT), a phylogenetically

    ancient signaling molecule (Hay-Schmidt, 2000), is the mostorks:opmental Disorders

    ience, ADHD Clinical Research Network, Department of Psychiatry,urzburg, Germanynce, Maastricht University, 6211 LK Maastricht, The Netherlands

    pment and modulates a wide spectrum of essentialitive appraisal to emotional responses in the matureundamentally impact the pathophysiology and long-understanding of how 5-HT-dependent modulationd emotional learning has been enhanced by recentsynapse formation and plasticity. In this review, weic plasticity impact our biosocial brain and how recentd function provide insight into the cellular and phys-

    states, interpersonal knowledge, and motivations as well as

    developmentally chiseled procedures used to decode and

    encode the self relative to other people. This complex set ofNeuron 76, October 4, 2012 2012 Elsevier Inc. 175

  • Neuronand plasticity, and network connectivity related to social cogni-

    tion and emotional learning. We explicitly focus on 5-HTs

    capacity to orchestrate activities and interactions of other trans-

    mitter systems by modifying the repertoire of molecules critically

    involved in the remodeling of transsynaptic signaling, high-

    lighting a selection of key players and newly discovered but

    paradigmatic mechanisms. This overview is not meant to be

    exhaustive but will touch upon emerging concepts of how defi-

    cits in 5-HT-moderated synaptic signaling contribute to the path-

    ophysiology of neurodevelopmental disorders.

    Structural and Functional SubsystemsThe mammalian brain 5-HT system originates from the raphe

    DR(B6, B7)HippocampusFrontal cortex

    Olfactory bulb

    MR(B5, B8)

    Hypothalamus

    ThalamusMFB

    Striatum

    N. accumbensAmygdala B9located in the midline of the rhombencephalon and in the retic-

    ular formation, where 5-HT neurons are clustered into nine nuclei

    numbered B1-9 on a rostrocaudal axis (Figure 1; Azmitia and

    Whitaker-Azmitia, 1997; Dahlstrom and Fuxe, 1964). These clus-

    ters are subdivided into rostral and caudal sections with the

    rostral subdivision comprising the caudal linear nucleus (CLi),

    the dorsal raphe nucleus (DR: B6, B7) and the median raphe

    nucleus (MR: B9, B8, and B5). 5-HT neurons from the rostral

    subdivision project primarily to the forebrain where the extensive

    collateralization of their terminals densely innervate virtually all

    regions (Calizo et al., 2011; Hensler, 2006; Hornung et al.,

    1990). A stringent topographical organization of two classes of

    fine and beaded fibers (termed D and M fibers, respectively)

    define distinct patterns of termination modulating specified

    arrays of neurons in the cortex, striatum, hippocampus, and

    amygdala (Figure 2), thus influencing sensory processing, cogni-

    tion, emotional states, circadian rhythms, food intake, and repro-

    duction. The caudal portion, which projects mainly to the spinal

    cord and cerebellum, consists of nuclei termed as raphe pallidus

    (B1), raphe obscurus (B2), and raphe magnus (B3) is involved in

    motor activity, pain control, and regulation of the autonomic

    nervous system. Here, the focus will be on the modulatory func-

    tion of the rostral subdivisions and the DR in particular.

    Based on cellular morphology, expression of other transmit-

    ters, afferent and efferent connections and functional properties,

    the circumscribed rap

    distinct 5-HT subsyste

    by transcriptional re

    factors that induce e

    the Lim homeodomai

    Lmx1b and Pet1, resp

    et al., 2011). Pet1 is on

    specification (Jacobse

    while Lmx1b represe

    expression cascade r

    of all 5-HT neurons i

    several secreted pos

    growth factors (Fgf4, F

    tically control cell fat

    (Cordes, 2005). Beyon

    action, the role of

    posttranscriptional rep

    tional regulation are in

    (see below).

    The 5-HT transporte

    display transient and

    development (Manso

    2001). For receptors, e

    expression patterns ar

    176 Neuron 76, October 4, 2012 2012 Elsevier Inc.., 2009; Lesch et al., 2012a). Even within

    he complex, morphogenetic programs in

    ms in rodents are differentially controlled

    gulators (Cordes, 2005). Transcription

    xpression of 5-HT markers encompass

    n and ETS domain transcription factor,

    ectively (Hendricks et al., 1999; Kiyasova

    e of the critical regulators of 5-HT system

    n et al., 2011; Liu and Deneris, 2011),Cerebellum

    MidbrainB1

    B2

    B3

    B4

    Figure 1. Rodent Brain 5-HT System5-HT neuron clusters are organized in the nineraphe nuclei, B1B9. The more caudal nuclei (B1B3) in the medulla project axons to the spinal cordand the periphery, whereas the more rostral raphenuclei contain the principal dorsal raphe group (B6and B7; depicted in yellow) and the median raphegroup (B5 and B8; depicted in green), whichproject to different but overlapping brain areas.DR, dorsal raphe nucleus; MFB, medial frontalbundle; MR, median raphe nucleus.

    5-HT neurons of the DR are topographi-

    cally grouped into six cell clusters

    comprising the rostral, ventral, dorsal,

    lateral, caudal, and interfascicular parts

    of the DR (Calizo et al., 2011; Hensler,

    2006; Waselus et al., 2006). In addition

    to 5-HT cells, neurons transmitting

    glutamate, GABA, dopamine, nitric oxide,

    and numerous neuropeptides (e.g., neuropeptide Y, galanin,

    somatostatin, thyrotropin-releasing hormone) were identified

    (Fu et al., 2010). Multiple brain regions feed back to the DR,

    utilizing a wide range of transmitters including glutamate, acetyl-

    choline, GABA, norepinephrine, or neuropeptides.

    Knowledge of themolecular mechanisms regulating the devel-

    opment of 5-HT system remains limited. The regulation of the

    proliferation, differentiation, maintenance and survival of 5-HT

    neurons engage many signaling molecules, including inducers

    of gene transcription, neurotrophic peptides, and steroids acting

    in concert or in cascade. Whether intrinsic neuronal, maternal or

    placental 5-HT is required as facilitator of 5-HT circuitry

    development remains controversial (Daubert and Condron,

    2010; Gutknecht et al

    Reviewnts a major determinant in the gene

    esulting in the phenotypic determination

    n brain (Song et al., 2011). Additionally,

    itional markers, including the fibroblast

    gf8) and Sonic hedgehog (Shh) synergis-

    e and the generation of 5-HT neurons

    d transcription initiation and neurotrophin

    mRNA elongation, microRNA-mediated

    ression and other mechanisms of transla-

    creasingly attracting systematic scrutiny

    r (5-HTT) and several 5-HT receptors also

    variable patterns of expression during

    ur-Robaey et al., 1998; Persico et al.,

    nzymes, and transporters, developmental

    e highly plastic, with prenatal exposure to

  • Neuron$

    Prefrontalcortex

    Dorsalstriatum

    TAN

    Review5-HT functionmodifying compounds or toxins causing long-term

    expression changes persisting into adulthood. Moreover,

    genetic variation in key players of 5-HT system development

    has been shown to affect the 5-HTs neuromodulatory capacity

    with consequences for the cognition-emotion continuum (Gross

    and Hen, 2004; Pessoa, 2008).

    Multiplicity of Signaling PathwaysSerotonergic input into neural networks implicated in sensory

    processing, cognitive control, emotion regulation, autonomic

    responses, and motor action is composed of two distinct 5-HT

    systems differing in their topographic organization, electrophys-

    iological signature, morphology, and sensitivity to neurotoxins

    and psychoactive compounds (Figure 2). There are at least

    fourteen structurally and pharmacologically divergent 5-HT

    receptors (Barnes and Sharp, 1999; Millan et al., 2008). Beyond

    isoform diversity, alternative splicing of some subtypes (e.g.,

    BLA

    LA

    Amygdala

    MSN

    Figure 2. Modulation of Glutamate- and GABA-Mediated TransmissionFor example, excitatory transmission within hippocampal areas CA13 is largely blayer II stellate cells in the entorhinal cortex, the mossy fiber axons originating fropyramidal cells. The synaptic communication of each of these pathways ismodulaand strength of the connection. 5-HT1B receptors, located on axon terminals fromto neighboring pyramidal neurons and to local interneurons. The release of GABA fand inhibited by 5-HT1A receptors. 5-HT and GABAB receptors, respectively, innosum moleculare. In the dentate gyrus, 5-HT3 receptors stimulate GABA releasemediated currents acting both pre- and postsynaptically; 5-HT and GABAB recSeveral 5-HT receptors are also expressed by cells of dorsal striatum includingneurons (TAN). In other brain regions distinct 5-HT receptors are required for difimplicated in regulating short-term plasticity. Presynaptically expressed 5-HT recand they are be distributed in a target-specific manner, such that synaptic input frits postsynaptic targets. 5-HT signaling specifies a mechanism for synaptic specithe strength and timing of network activity within pyramidal cells, other principleDorsalhippocampus

    CA1

    DG

    5-HT projections

    Thin, varicose axon system (D fibers)Basket axon system (M fibers)5-HT4) and RNA editing of the 5-HT2C receptor add to the diver-

    sity of the 5-HT receptor family. It continues to be a daunting task

    to dissect the physiological impact of individual receptors,

    design selective ligands to target specific subtypes, and deter-

    mine potential therapeutic value of novel compounds. Molecular

    characterization of 5-HT receptor subtypes, functional mapping

    of transcriptional control regions, and the modeling of 5-HT

    receptor gene function in genetically modified mice has yielded

    valuable information regarding respective roles of 5-HT recep-

    tors and other components of serotonergic signaling pathways

    in brain development, synaptic plasticity, and behavior.

    The well-characterized 5-HT1A subtype is a G protein-

    coupled receptor (GCPR) that operates both pre- and postsyn-

    aptically (Figure 2). Somatodendritic 5-HT1A autoreceptors are

    predominantly located on the soma and dendrites of neurons

    in the raphe complex and its activation by 5-HT or 5-HT1A

    agonists induces hyperpolarization, decreases the firing rate of

    DR

    MR

    CA3

    PyramidalPV

    GABAergic neurons neuron Non-PV

    Midbrain5-HT1A

    5-HT35-HT2C5-HT2A5-HT1B

    5-HT4

    by 5-HT in the Cortex, Striatum, Hippocampus, and Amygdalaased on three glutamatergic pathways: the perforant path formed by axons ofm the dentate gyrus granule cells, and the recurrent axon collaterals of CA13ted by 5-HT receptors that fine-tune synaptic signal by affecting both the timingpyramidal neurons and on their recurrent collaterals, inhibit glutamate release

    romCA1 inhibitory interneurons is stimulated by 5-HT2 and by 5-HT3 receptorscrease and decrease T-type Ca2+ current on interneurons from stratum lacu-from interneurons. In CA3 pyramidal neurons, 5-HT inhibits GABAB receptor-eptors cooperate in increasing a hyperpolarizing outward potassium current.the medium-sized spiny neurons (MSN), the tonically active cholinergic inter-ferent forms of pre- and postsynaptic long-term plasticity and also have beeneptors affect the timing of action potentials elicited in the postsynaptic targetom one presynaptic neuron can be modulated by different receptors at each ofalization of glutamatergic and GABAergic transmission and thus contributes toneurons and interneurons.

    Neuron 76, October 4, 2012 2012 Elsevier Inc. 177

  • Neuron5-HT neurons, and subsequently reduces the synthesis, turn-

    over, and release of 5-HT from axon terminals in projection areas

    (Gutknecht et al., 2012; Lesch, 2005). Postsynaptic 5-HT1A

    receptors are widely distributed in forebrain regions, notably in

    the cortex, hippocampus, septum, amygdala, and hypothal-

    amus. Hippocampal heteroreceptorsmediate neuronal inhibition

    by coupling to the G protein-gated inward rectifying potassium

    channel subunit-2 (GIRK2). The metabotropic and ion channel-

    regulating actions of the 5-HT1A receptor are implicated in

    learning and memory (Ogren et al., 2008) and in the pathophys-

    iology and treatment response of a wide range of disorders

    characterized by cognitive and emotional dysregulation (Gross

    and Hen, 2004; Gross et al., 2002). Chronic stress mediated by

    glucocorticoids has been reported to result in downregulation

    of 5-HT1A receptors in the hippocampus in animal models

    (Meijer et al., 1998). In line with this notion, evidence is accumu-

    lating that functional variation in the 5-HT1A gene (HTR1A) is

    associated with personality traits of negative emotionality (Stro-

    bel et al., 2003) as well as the etiology of disorders of the anxio-

    depressive spectrum (Rothe et al., 2004; for review, Albert, 2012;

    Le Francois et al., 2008).

    By associating with multiple GPCR interacting proteins, two

    other metabotropic receptors, 5-HT2A and 5-HT2C, impact

    a wide range of signal transduction pathways (Bockaert et al.,

    2010). Both 5-HT2A and 5-HT2C receptor-agonist complexes

    activate phospholipase C (PLC). 5-HT2C is critically involved in

    the regulation of synaptic plasticity, since it initiates the phos-

    phoinositol second messenger cascade by producing inositol

    triphosphate (IP3) and diacylglycerol (DAG), which ultimately

    leads to opening L-type Ca2+ channels following release of

    calcium stores. Moreover, the protein phosphatase and tensin

    homolog (PTEN) binds to 5-HT2C, and disruption of 5-HT2C/

    PTEN complexes can alter neuronal activity (Bockaert et al.,

    2010). There is evidence that the 5-HT2C receptor also interacts

    with proteins containing PSD-95-disc large-zonula occludens

    (PDZ) domains, and association of 5-HT2C receptors with PDZ

    proteins affects both receptor desensitization and internaliza-

    tion, depending on the type of the PDZ protein associated with

    the receptor (Becamel et al., 2004). The spatiotemporal diversity

    of these interactions highlights the wide range of 5-HT-mediated

    adaptive plasticity at the synaptic level.

    5-HT1A and 5-HT2A/2C receptors can be expressed in

    both excitatory principal neurons and inhibitory interneurons

    (Figure 2), which renders the net outcome of the neuromodula-

    tory action of 5-HT on circuit activity dependent on multiple

    factors (e.g., local 5-HT concentration, receptor ratio and intra-

    cellular coupling) (Cruz et al., 2004; de Almeida and Mengod,

    2008; Llado-Pelfort et al., 2012; Puig et al., 2005). On glutamater-

    gic pyramidal neurons, 5-HT1A receptors are distributed

    diffusely and at relatively high density over the perikaryon,

    dendrites, and synaptic spines, whereas 5-HT2A/2C receptors

    are localized to the proximal dendritic shafts of glutamatergic

    pyramidal neurons, and more diffusely on synaptic spines, inclose association with glutamate receptors (de Almeida and

    Mengod, 2007; Gonzalez-Maeso et al., 2008). In addition, 5-

    HT1A and 5-HT2A/2C receptors are found on terminals and

    perikarya of GABAergic interneurons, respectively (de Almeida

    and Mengod, 2008; Navailles and De Deurwaerdere, 2011).

    178 Neuron 76, October 4, 2012 2012 Elsevier Inc.5-HT1A activation decreases N-methyl D-aspartate (NMDA)

    receptor-mediated currents in pyramidal neurons of the pre-

    frontal cortex (PFC) through reduction of ERK1/2 activity, which

    leads to a decrease in microtubule-associated protein-2 (MAP2)

    phosphorylation, MAP2-microtubule interaction and microtu-

    bule stability involved in clustering the NMDA receptor-2B

    subunit (Yuen et al., 2005). In contrast, 5-HT2A/2C activation

    increases NMDA receptor-mediated currents by activating the

    ERK1/2 pathway via the b-arrestin/Src/dynamin cascade, thus

    counteracting the effects of 5-HT1A activation in decreasing

    NMDA receptor-mediated currents (Yuen et al., 2008). Thus,

    5-HT1A- and 5-HT2A/2C-activated signaling pathways appear

    to converge at antagonistic actions on ERK1/2.

    Somatosensory Cortex DevelopmentConverging lines of evidence suggest differential roles of 5-HT in

    the developing and adult brain. During specific time windows of

    embryogenesis, 5-HT in concert with other transmitters regu-

    lates brain cytoarchitecture and nodal connectivity by modu-

    lating a wide variety of developmental processes, including

    neural progenitor cell proliferation, migration and differentiation,

    maturation of postmitotic neurons and apoptosis (Erzurumlu and

    Gaspar, 2012, and references therein). Environmental factors

    that alter serotonergic modulation during development or varia-

    tion in genes involved in 5-HT signaling can cause disorders

    associated with defective innervation, circuit formation, and

    network connectivity.

    Numerous investigations of 5-HTs participation in neocortical

    development and plasticity focused on the rodent visual and

    particularly the somatosensory cortex (SSC), due to its one-to-

    one correspondence between the sensory system and its

    cortical projection area (Figure 3). Here, to provide an example

    of how the serotonergic system can impact cortical develop-

    ment, we consider the formation of the SSC and its activity-

    dependent plasticity. The pronounced growth of the cortex

    during development coincides with progressive serotonergic

    innervation. During this period, incoming 5-HT neuron terminals

    begin to establish synaptic interactions with target neurons and

    to elaborate a profuse branching pattern, matching the transient

    barrel-like expression and distribution of 5-HT, 5-HT1B, and

    5-HT2A receptors as well as the 5-HTT, which regulates extra-

    cellular 5-HT levels by mediating high-affinity reuptake, in

    early-postnatal primary SSC (Mansour-Robaey et al., 1998).

    The barrel-like 5-HT pattern in layer 4 of the SSC stems from

    5-HT uptake and vesicular storage in thalamocortical neurons,

    transiently expressing both 5-HTT and the vesicular monoamine

    transporter-2 (VMAT2) despite their ultimate glutamatergic

    specification.

    5-HT dysregulation profoundly disturbs formation of the SSC

    with altered cytoarchitecture of cortical layer 4, the layer

    that contains synapses between thalamocortical terminals and

    their postsynaptic target neurons (Persico et al., 2001). 5-Htt

    knockout mice display a lack of characteristic barrel-like clus-

    Reviewtering of layer 4 neurons in the SSC, despite relatively preserved

    trigeminal and thalamic patterns (other phenotypes of 5-Htt-

    deficient mice are described in Figure 4). 5-HT synthesis inhibi-

    tion within a narrow early postnatal time window (P0P4)

    completely rescues formation of SSC barrel fields, indicating

  • 123

    4

    5

    6

    123

    4

    5

    6

    GLU

    5-HTRaphe

    NMDAAMPA

    5-HT2A

    TCA

    GLU

    5-HT1B

    5-HTT

    GLU

    5-HTRaphe

    AMPA

    5-HT2A

    5-HTTAMPA

    TCA

    GLU

    GLU

    5-HTRaphe

    NMDAAMPA

    5-HT2A

    TCA

    GLU

    5-HT1B 5-HT5-HT 5-HT

    Adult cross-modalsynaptic plasticity

    S1 cortex

    5-HTneuronterminals

    5-HT transporterinactivation

    LTP

    Layer 4barrel

    123

    4

    5

    6

    CA B

    Barrel cortex

    Neonataldevelopment

    GanglionGasseri

    V sensory nerveth

    Ventrobasalthalamus

    Figure 3. Development and Plasticity of the Somatosensory Cortex(A) The rodent somatosensory cortex (SSC) is characterized by one-to-one correspondence between the sensory system and its cortical projection area. Eachwhisker on the rodent snout is somatotopically represented in the trigeminal nucleus (termed barrelette), ventro-postero-medial thalamus (barreloid) and primarysomatosensory cortex (barrel). Cortical barrels encompass a hollow center with abundant thalamocortical terminals and few granule cells in layer 4, surroundedby a ring of dense granule cells separated by septal spaces. Thalamocortical afferents (TCA) from the ventrobasal thalamic nucleus are distributed somato-topically perinatally and play an instructive role in subsequent cortical barrel field formation. Afferents-instructed barrel formation is representative of theperipheral-to-central maturation cascade, with barrelettes forming prenatally, barreloids approximately at birth and barrels around P4. Peripheral sensory input,e.g., via whisker-mediated stimuli, is critical to the organization of the barrel field during an early postnatal critical period (i.e., P0P4).(B) 5-Htt knockout mice display a lack of characteristic barrel-like clustering of layer 4 neurons in the SSC, despite relatively preserved trigeminal and thalamicpatterns. Cell bodies as well as terminals, typically more dense in barrel septa, appear homogeneously distributed in layer 4 of adult brains. Excessiveconcentrations of extracellular 5-HT are deleterious to SSC development suggesting that transient 5-HTT expression and its permissive action in thalamocorticalneurons is required for normal barrel pattern formation in neonatal rodents, by maintaining extracellular 5-HT concentrations below a critical threshold. 5-HT1Breceptors are the direct targets of excess 5-HT.While activation of 5-HT1B inhibits neurotransmitter release, specifically reducing excitatory neurotransmission inthalamocortical regions of somatosensory systems, 5-HT1B receptors act as regulators of thalamocortical development through inhibition of glutamate (GLU)release. Since normal synaptic density of 5-HT neuron terminal in SSC layer 4 of 5-Htt knockout mice is maintained, it is likely that 5-HT affects SSC cy-toarchitecture by promoting dendritic growth toward the barrel hollows, as well as by modulating cytokinetic movements of cortical granule cells.(C) 5-HT also moderates activity-dependent cross-modal plasticity, a procedure of cortical restructuring to compensate for the loss of one sensory systemwith other intact modalities in the mature brain, specifically among the SSC and visual system. Increases in extracellular 5-HT in the rodent SSC followingvisual deprivation enables synaptic strengthening at layer 4 to layer 2/3 synapses in response to whisker-dependent stimulation of neural activity. Theenhanced transsynaptic signaling efficiency is achieved by insertion of AMPA receptors into synapses at postsynaptic neurons through activation of the5-HT2A/2C-dependent ERK1/2 signaling pathways and increased phosphorylation of AMPA receptor subunit GluR1, thus leading to sharpening andfine-tuning of the functional whisker-barrel map at layer 4-2/3 at an age when natural whisker experience fails to induce synaptic GluR1 delivery. LTP, long-termpotentiation.

    Neuron 76, October 4, 2012 2012 Elsevier Inc. 179

    Neuron

    Review

  • incth

    Neuronet al., 2010) and that intra- and extra-uterinematernal signals can synergisticallyand psychiatric disorders (Bartolomucci et al., 2010; Carola et al., 2008; Leshaploinsufficiency of 5-HT system interacting genes, such as Bdnf or Pten, furthat excessive concentrations of extracellular 5-HT are delete-

    rious to SSC development. Thus, by maintaining extracellular

    5-HT concentrations below a critical threshold, transient 5-HTT

    expression and its permissive action in thalamocortical neurons

    is required for normal barrel pattern formation in neonatal

    rodents. Converging lines of evidence support 5-HTB receptors

    as direct targets of excess 5-HT. Since activation of 5-HT1B

    inhibits transmitter release, specifically reducing excitatory

    transmission in thalamocortical regions of both the visual and

    somatosensory systems, hypotheses based on modulation

    of electrophysiological activity view 5-HT1B receptors as

    regulators of thalamocortical development through inhibition of

    glutamate release (Salichon et al., 2001). Since normal synaptic

    density of 5-HT neuron terminal in SSC layer 4 of 5-Htt knockout

    mice ismaintained, it is likely that 5-HT affects SSC cytoarchitec-

    ture by promoting dendritic growth toward the barrel hollows, as

    well as by modulating cytokinetic movements of cortical granule

    cells. In total, the interplay of 5-HT synthesis, release, uptake and

    degradation by raphe-cortical and thalamocortical axon arbors

    at target neurons and subsequent differential activation of me-

    tabotropic 5-HT1B receptors plays a critical role in the formation

    of sensory and potentially other cortical fields.

    Activity-Dependent Cortical PlasticityNeuronal plasticity in the mature cortex is regulated by cognitive

    and emotional functions such as processes related to percep-

    tion, attention, motivation, associative and emotional learning,

    and memory (Holtmaat and Svoboda, 2009). By innervating

    regions implicated in higher-order brain function, the 5-HT

    sociability in a sex-specific manner (Page et al., 2009; Ren-Patterson et al., 20behavior underscores the view that environmental influences can persistently reminformative for the dissection of the molecular and neural mechanisms of epiginteractions may constitute neural mechanisms for (epi)genetic vulnerability towa

    180 Neuron 76, October 4, 2012 2012 Elsevier Inc.Figure 4. 5-HT Transporter as a MasterController of Brain 5-HT System FunctionTargeted mutations resulting in reduced orcompletely inactivated 5-Htt (Sert, Slc6a4) func-tion in mice have led to the identification of morethan 50 different phenotypic changes, rangingfrom increased anxiety and stress-related behav-iors to gut dysfunction, bone weakness, and late-onset obesity with metabolic syndrome (Murphyand Lesch, 2008). These multiple effects, whichmay be amplified by gene-by-environment (G3E)and gene-by-gene (G3G) interactions, are pri-marily attributable to altered intracellular andextracellular 5-HT concentrations during sensitiveperiods in development and adulthood (Ansorgeet al., 2004). In addition, the 5-Htt knockout mouseprovides a model to study the impact of geneticmechanisms on development and plasticity ofthe brain including regionalization of the cortexand its connectivity to subcortical structures(Nietzer et al., 2011; Persico et al., 2001; Salichonet al., 2001; Wellman et al., 2007). Furthermore,evidence indicates that 5-HT-dependent tran-scriptional programming of maternal behavior haslong-lasting consequences on (social) cognitiveand emotional behavior in the offspring (Barr et al.,2004; Bennett et al., 2002; Canli et al., 2006;Jansen et al., 2011; Kloke et al., 2011; Lewejohann

    duce enduring plastic changes in neurocircuits involved in neurodevelopmentalh, 2011; van den Hove et al., 2011). Inactivation of 5-Htt in interaction wither aggravates deficits in brain development as well as emotional behavior and

    Reviewsystem plays a predominant role in the modulation of these

    functions. Although dynamic cortical reorganization of areas

    involved in cognition and emotion is critical for this adaptation

    and the enhancement of neural plasticity in response to activa-

    tion of the raphe 5-HT system is well established (Bennett-Clarke

    et al., 1996; Inaba et al., 2009; Jones et al., 2009; Kim et al., 2006;

    Maya Vetencourt et al., 2008; Normann and Clark, 2005), the

    underlying molecular, synaptic, and circuit mechanisms are

    only beginning to be adequately understood.

    Raphe 5-HT neurons orchestrate cortical reorganization

    among different sensory and effector systems via modification

    of transsynaptic signaling efficiency at excitatory synapses. In

    the mammalian brain, the majority of excitatory synapses use

    glutamate as transmitter. Glutamate activates both ionotropic

    (AMPA-, kainate-, and NMDA-type) receptors and metabotropic

    (mGluR) receptors. Fast glutamatergic transmission is primarily

    mediated by AMPA receptors, while mGluRs modulate the

    response to ionotropic glutamate receptors and that of other

    transmitters, including dopamine, 5-HT, and GABA (De Blasi

    et al., 2001). The principal cellular mechanism for 5-HT to impact

    synaptic plasticity is long-term potentiation (LTP), an enduring

    increase in synaptic transmission efficiency that has been

    proposed to represent the physiological basis of learning and

    memory. Synaptic delivery and insertion of AMPA receptors

    mediated by lateral diffusion from extrasynaptic sites appears

    central to the induction of postsynaptic LTP (Bredt and Nicoll,

    2003; Malinow and Malenka, 2002; Figure 5). Detailed knowl-

    edge about the molecular mechanisms underlying 5-HT-medi-

    ated plasticity is now emerging and it has become clear that

    06; Ren-Patterson et al., 2005). This (epi)genetic programming of emotionalodel neuronal units during early development, rendering 5-Htt modified miceenetic programming at the neurodevelopmental-behavioral interface. Theserd, or resilience against disease.

  • Neuronserotonergic signaling modulates intracellular pathways in-

    volved in synaptic AMPA receptor delivery. Activation of the

    5-HT2A-dependent ERK1/2 pathways enhances transsynaptic

    signaling efficiency via insertion of AMPA receptors into

    synapses at postsynaptic neurons (Makino and Malinow, 2009)

    and increased phosphorylation of AMPA receptor subunit

    GluR1 at Ser845 (Derkach et al., 2007).

    5-HT also moderates cross-modal plasticity, a procedure of

    cortical restructuring to compensate for the loss of one sensory

    system with other intact modalities in the mature brain, specifi-

    cally among the SSC and visual system. Increases in extracel-

    lular 5-HT in exclusively the rodent barrel cortex following visual

    deprivation enables synaptic strengthening at layer 4 to layer 2/3

    synapses in response to whisker-dependent stimulation of

    neural activity (Jitsuki et al., 2011). The enhanced transsynaptic

    signaling efficiency due to AMPA receptor addition to synapses

    leads to sharpening and fine-tuning of the functional whisker-

    barrel map at layer 4-2/3 at an age when natural whisker experi-

    ence fails to induces synaptic GluR1 delivery. Taken together,

    sensory deprivation of onemodality increases 5-HT release in re-

    maining modalities, which in turn modulates intracellular

    signaling pathways involved in AMPA receptor delivery facili-

    tates GluR1-subunit dependent synaptic strengthening, and

    enables cortical reorganization, thus improving whisker barrel-

    dependent sensory function.

    Powering Synaptic Plasticity: Molecules for Bridging theCleftWhile enhancement of plasticity in response to activation of the

    5-HT system has been well-established by electrophysiological

    approaches, the underlying molecular mechanisms are now un-

    folding. Synaptic adhesion molecules and secreted signaling

    proteins regulate distinct aspects of neuronal circuitry formation

    and function. Coordinated actions of a large diversity of molec-

    ular signals contribute to the specification and differentiation of

    synaptic connections in the developing and mature brain.

    Evidence has been accumulating that 5-HT signaling modulates

    these adhesion complexes. In this section, we provide a brief

    overview of synaptic adhesion molecules and their functions.

    Establishment of functional circuits and tight regulation of

    connectivity require precision and specificity of neural wiring at

    the laminar, cellular, subcellular, and synaptic levels (Williams

    et al., 2010a). Transmembrane adhesion proteins are essential

    constituents of synapses that play fundamental roles in building

    and maintaining synaptic structure during development and

    serve diverse purposes in synaptic plasticity of the brain

    throughout the entire life span (Benson et al., 2000; Dalva

    et al., 2007; Murase and Schuman, 1999; Yamagata et al.,

    2003). There is a wide diversity of synaptic adhesion molecules

    and here we discuss only those that have been identified at the

    crossroads of 5-HT-dependent synaptic plasticity and the path-

    ogenesis of neurodevelopmental disorders. These include integ-

    rins, immunoglobulin superfamily (e.g., neural cell adhesion

    Reviewmolecules, NCAMs), cadherins (CDHs), adhesion G protein-

    coupled receptors (adhesion-GPCR; e.g., latrophilins, LPHNs),

    leucine-rich repeat transmembrane proteins (e.g., LRRTMs,

    FLRTs), neurexins (NRXNs) ,and neuroligins (NLGNs) (de Wit

    et al., 2009; OSullivan et al., 2012; Sudhof, 2008; Williamset al., 2010a, 2011; Figure 5). A diversity of synaptic adhesion

    molecules, including, e.g., NCAM1, NRXN1 and 3, CDH8, 11,

    and 13, LPHN1 and 3, are expressed by serotonergic neurons

    and some are subject to transcriptional regulation during the

    process of synapse formation and remodeling (Bethea and

    Reddy, 2012a, 2012b; Lesch et al., 2012b; Rivero et al., 2012;

    Wylie et al., 2010).

    Adhesion molecules modulate synapse formation by speci-

    fying the connectivity between matched populations of neurons.

    Once the synaptic partner is identified, the initial axo-dendritic

    contact is transformed into a functional synapse by the recruit-

    ment of other pre- and postsynaptic components. A well-char-

    acterized mediator of synaptogenesis is the transsynaptic

    NRXN-NLGN complex, in which presynaptic NRXNs interact

    with postsynaptic NLGNs to bidirectionally specify synapses

    (Sudhof, 2008). Although all neurons express NRXNs and

    NLGNs, alternate promoter usage and extensive alternative

    splicing of extracellular domain generates numerous different

    isoforms of NRXNs likely confering specificity for glutamatergic

    versus GABAergic synapse formation. Although NRXNs,

    NLGNs, and LPHNs are structurally distinct, they display hetero-

    philic interaction between their extracellular domains (Boucard

    et al., 2012). By specifying synaptic functions, multiple parallel

    transsynaptic signaling complexes shape unique network prop-

    erties (Benson et al., 2000; Bockaert et al., 2010).

    Synaptic adhesion molecules share the ability to trigger

    multiple intracellular signaling cascades with metabotropic

    5-HT and glutamate receptors as well as neurotrophin receptors

    (Figure 5). The cytoplasmic domain of both NRXNs and NLGNs

    contains PDZ-binding motifs that recruit messenger molecules

    thought to mediate differentiation of the presynaptic and the

    postsynaptic compartment, respectively. Several intracellular

    signaling pathways may be activated by LPHNs via both Ca2+-

    dependent and -independent mechanisms. The Ca2+-indepen-

    dent effects are likely transduced by G proteins that trigger acti-

    vation of both PLC and inositol-3-phosphate (IP3), resulting

    in Ca2+ mobilization from intracellular Ca2+ stores, eventually

    followed by release of neurotransmitters. Moreover, LPHNs

    C-terminal regions interact with proteins of the SHANK family

    (Kreienkamp et al., 2000), multidomain scaffold proteins of the

    postsynaptic density that connect neurotransmitter receptors,

    ion channels, and other membrane proteins to the actin cyto-

    skeleton and G protein-coupled signaling pathways and also

    play a role in synapse formation and dendritic spine maturation

    (Holtmaat and Svoboda, 2009).

    At the glutamatergic postsynapse, SHANKs couple to the

    NMDARPSD-95nitric oxide synthase-1 (NOS1) complex via

    interaction with the GKAP adaptor protein (Naisbitt et al., 1999;

    Romorini et al., 2004). While GKAP is thought to be a PSD-95

    associated scaffolding protein maintaining synaptic junctions

    and synaptic stability, the PSD complex also operates as a func-

    tional link as it tightly couples the NMDA receptor to NOS1. The

    latter is able to bind to PSD-95 by a unique PDZ-PDZ domaininteraction, allowing for attachment of NOS1 to the NMDA

    receptor complex. NOS1, which has also been reported to recip-

    rocally interact with 5-HTT function (Chanrion et al., 2007), is

    spatially close to where Ca2+ influx occurs, which activates

    NOS1. Lastly, SHANKs bind to HOMER proteins, another group

    Neuron 76, October 4, 2012 2012 Elsevier Inc. 181

  • Glutamate

    MINT-1 Ca2+channel

    PI3KERK1/2AKTGSK3mTOR

    mGLUR5

    Ribosome

    F-actin

    NMDAR

    PSD-95

    NLG

    N

    SHANK GKAP

    HOMER

    RhoA/ROCKRAC1

    VELICASK

    MTRRL

    NOS1

    PKC

    PTENPSD-95MPP3

    CaMK

    IP3

    [Ca2+]Synapticproteins

    SAP-97

    NXRN

    NLG

    N

    mGLUR75-HT1B

    5-HT2A/2C

    Serotonin

    NXRN

    AMPAR5-HTT

    DAG

    5-HT1BP11

    F-actin

    Ribosome

    Ribosome

    AC

    PKC

    cAMP

    PLCIP3

    [Ca2+]DAG

    Gq

    FGFRNCAMPLC

    F-actin

    PICK1

    PKASynapticproteins

    Gi

    PKA

    GIRK2

    PKA

    GSK3

    ( e.g. glutamate,GABA, dopamine,acetylcholine )

    5-HT1A

    PI3KERK1/2AKTGSK3mTOR

    CDH

    Spine volume

    PSD

    size

    N

    MD

    AR/

    AM

    PAR

    ratio LTP

    LTD

    Synapticproteins

    NHPL

    5-HT1A

    Figure 5. Prototypical Glutamatergic Synapse Modulated by Serotonergic InputThe model character of this hypothetical synapse allows depiction of multiple receptors and their postreceptor signal transduction, transsynaptic interactionsof adhesion molecules, and related intracellular signaling pathways regulating synaptic plasticity in a single schematic representation. For simplificationpurposes only a fraction of 5-HT receptor subtypes is depicted. Somatodendritic (5-HT1A) and terminal autoreceptors (5-HT1B) induce hyperpolarization thatdecreases the firing rate of 5-HT neurons. 5-HT2A and 5-HT2C basically activate phospholipase C (PLC) via the Gq protein but also impact a wide range ofdistinct signal transduction pathways by associating with multiple GPCR interacting proteins. The 5-HT2C stimulates the phosphoinositol second messengercascade which, in turn, stimulates the activation of protein kinase C (PKC) and opens L-type Ca2+ channels. The protein phosphatase and tensin homolog(PTEN) binds to the 5-HT2C receptor which also interacts with several proteins, such as postsynaptic density (PSD) proteins containing PSD-95-disc large-zonula occludens (PDZ) domains. Association of 5-HT2C receptors with PDZ domain-containing proteins affects both receptor desensitization and inter-nalization, depending on the type of the PDZ protein associated with the receptor. Neurexins (NRXNs) and their postsynaptic binding partners neuroligins(NLGNs), leucine-rich repeat transmembrane proteins (e.g., LRRTMs, FLRTs) and adhesion G protein-coupled receptors (adhesion-GPCR; e.g., latrophilins,LPHNs). Other synaptic adhesion proteins are members of the immunoglobulin superfamily (e.g., neural cell adhesion molecules, NCAMs) or cadherin family(e.g., CDH9, atypical CDH13). NRXNs interact with the scaffolding molecule CASK and NLGNs interact with the scaffolding molecule PSD-95 or SAP-97,which binds NMDA and AMPA receptors (NMDAR and AMPAR) via their PDZ domain. Alternatively spliced NRXNs bind the postsynaptic adhesion moleculeLRRTM2, which can recruit NMDARs and AMPARs. NCAMs interact with the fibroblast growth factor receptor (FGFR) signaling (PI3K, ERK, AKT, GSK3,mTOR) pathway, which is also activated by CDHs. Several intracellular signaling pathways may be activated by LPHNs including Ca2+-dependent and-independent mechanisms. Moreover, LPHNs C-terminal region interacts with proteins of the SHANK family. SHANK proteins are synaptic multidomain PSDscaffold proteins binding to HOMER proteins, another group of postsynaptic density scaffolding proteins, which, in turn, are able to interact with mGLUR5.SHANK and HOMER proteins cross-link mGLURs with LPHNs. 5-HT1A activation decreases NMDA receptor-mediated currents in PFC pyramidal neuronsthrough reduction of ERK1/2 activity, which leads to a decrease in microtubule-associated protein-2 (MAP2) phosphorylation, MAP2-microtubule interactionand microtubules stability involved in clustering the NMDA 2B subunit. In contrast, 5-HT2A/2C activation increases NMDA receptor-mediated currents byactivating the ERK1/2 pathway, thus counteracting the effects of 5-HT1A activation in decreasing NMDA receptor-mediated currents. Metabotropic glutamatereceptors (e.g., mGluR5, mGluR7) stimulate protein kinase A (PKA) and PKC pathways. mGluR5 not only interacts with signaling of 5-HT receptors but alsowith NMDA receptors resulting in reciprocal and agonist-independent inhibition of the two receptors. P11 and GSK3 may directly interact with 5-HT1B,

    182 Neuron 76, October 4, 2012 2012 Elsevier Inc.

    Neuron

    Review

  • roaohehetr

    Neuron

    Reviewof postsynaptic density scaffolding proteins (Tu et al., 1999; Xiao

    et al., 2000), which, in turn, are able to interact with mGluR1 and

    nuclei to various association cortices, while physiological responses can be pthe hypothalamus and brainstem. Excessive or insufficient activation of thesensitivity to social signals. The orbitofrontal cortex, through its connectionswithin limiting emotional outbursts, and the anterior cingulate cortex recruits otenvironmental factors contribute to the structure and function of this circuitry, tassociate stimuli with events that are either punishing or rewarding. vmPFC, venarea; LC, locus ceruleus.mGluR5. SHANK and HOMER proteins can cross-link mGluRs

    with LPHN3, which hence, in addition to its interaction with

    FLRT3 and subsequent G protein signaling, impacts glutamater-

    gic transmission in a dual mode (OSullivan et al., 2012). The

    signaling pathway activating interaction of synaptic adhesion

    molecules ultimately converges on the machinery regulating

    gene transcription which, in turn, results in de novo synthesis

    of structural and functional synaptic proteins by local ribosomes.

    Serotonin-Induced Modulation of Adhesion MoleculesFacilitates Remodeling of Synapses in EmotionalLearningAs a prototypical network subject to 5-HT-induced modulation,

    the circuitry of experience-dependent associative and emotional

    learning has been implicated in social cognition and emotion,

    including the associated phenomena of contextual fear re-

    sponses (Figure 6; LeDoux, 2012). While a complex develop-

    mental program encodes the formation and function of this

    circuitry, the amygdala governs essential processes ranging

    from cognition to emotion, to learning and memory (Phelps,

    2006). While genetic variation and environmental factors

    contribute to the structure and function of this circuitry, the

    amygdala-associated network is centrally involved in processes

    of learning to associate stimuli with events that are either punish-

    ing or rewarding, commonly referred to as emotional learning.

    whereas PTEN, MPP3, and PSD-95 are able to bind to 5-HT2C. Long-term potePSD size, and NMDR/AMPAR ratio are synaptic plasticity events that increase orLTP requires increased gene transcription and de novo synthesis of structurasupport the molecular basis of learning and memory.The recognition of the amygdala as an essential neural substrate

    for acquisition and expression of learned fear has permitted

    Figure 6. Serotonin and the Neurocircuitryof Emotional LearningModified from Lesch (2007). A network of inter-connected structures modulated by the median(MR) and dorsal (DR) raphe 5-HT system has beenimplicated in emotion learning, specifically fearresponse. Fear-related circuits involve pathwaystransmitting information to and from the amygdalato various neural networks that control theexpression of avoidant, defensive, or aggressivereactions, including behavioral, autonomic, andstress hormone responses. While pathways fromthe thalamus and cortex (sensory and prefrontal)project to the amygdala, inputs are processedwithin intra-amygdalar circuitries and outputs aredirected to the hippocampus, brain stem, hypo-thalamus, and other regions. Perception of dangeror threat and other social cues are transmittedto the lateral nucleus of the amygdala, whichprojects to the basal nuclei where informationregarding the social context derived from orbito-frontal projections is integrated with the percep-tual information. Behavioral responses can then beinitiated via activation of projections from the basal

    duced via projections from the basal nuclei to the central nucleus and then tomygdala leads to either disproportionate negative emotionality or impairedther domains of the prefrontal cortex andwith the amygdala, plays a critical roler entities during incremental levels of arousal. While genetic variation andamygdala-associated network is centrally involved in processes of learning toomedial prefrontal cortex; SSC, somatosensory cortex; VTA, ventral tegmentalelectrophysiological characterization of synaptic processes in

    the amygdala that mediate fear conditioning. Although the

    mechanisms underlying the induction and expression of LTP in

    the amygdala are only beginning to be understood, LTP induces

    postsynaptic GluR1 delivery in amygdala in conjunction with

    modified presynaptic plasticity in the lateral nucleus (Maren,

    2005; Rumpel et al., 2005). Reduction of NLGN-1 expression in

    pyramidal neurons of the lateral amygdala decreases NMDAR-

    dependent postsynaptic currents, impairing LTP at thalamo-

    amygdalar synapses, and triggers deficits in conditioned fear

    memory storage, consistent with the requirement of NMDA

    receptor activation for expression of synaptic plasticity in mature

    neural circuits in the amygdala (Kim et al., 2008).

    An impact of 5-HT on NRXN and NLGN function at sensory-to-

    motor neuron synapses of the gill-withdrawal reflex in Aplysia,

    which exhibits sensitization, named long-term facilitation (LTF),

    as a basic mechanism of conditioned fear response, was

    recently demonstrated (Choi et al., 2011). LTF is induced through

    5-HT receptor-mediated activation of cAMP-dependent PKA

    or PKC. These effectors subsequently recruit the mitogen-

    activated kinase (MAPK) signaling pathway which in turn initiates

    transcription factor CREB-dependent modulation of transcrip-

    tional activity. Suppression of NRXN in the presynaptic sensory

    neuron or NLGN in the postsynaptic motor neuron eliminates

    both LTF and the associated presynaptic growth provoked by

    ntiation (LTP) and long-term depression (LTD) as a function of spine volume,decrease the strength of synaptic transmission for long periods, respectively.l and functional synaptic proteins by local ribosomes which are believed to

    Neuron 76, October 4, 2012 2012 Elsevier Inc. 183

  • Neuronrepetitive application of 5-HT. Moreover, introduction of an

    autism-linked NLGN-3 mutation into the postsynaptic motor

    neuron decreases transsynaptic signaling efficiency reflected

    by obliteration of LTF. The maintenance of LTF and synaptic

    growth requires ribosome-mediated synaptic protein synthesis

    and is dependent on the translational regulator, cytoplasmic pol-

    yadenylation element-binding protein (CPEB) (Miniaci et al.,

    2008; Si et al., 2003). The findings further support the notion

    that 5-HT-induced recruitment of NRXNs and NLGNs partici-

    pates in the different stages of emotional memory formation

    and to learning-related structural remodeling that results in an

    expansion of synaptic connections and increase in signaling effi-

    ciency associated with storage of long-term memory, including

    emotional memory. Thus, 5-HT-evoked moderation of activity-

    dependent regulation of NRXN-NLGN interaction likely governs

    transsynaptic signaling required for the cognitive and emotional

    processes that are impaired in neurodevelopmental disorder.

    Serotonin-Moderated Epigenetic Programming ImpactsSynaptic PlasticityEnvironmental adversity and early-life stress experience during

    gestation and the postnatal period are associated with increased

    risk for neurodevelopmental disorders and psychiatric condi-

    tions later in life. A considerable number of human and animal

    model studies indicate that the impact of gene-by-environment

    interaction on brain development and functionspecifically in

    the domain of social cognition and emotional learningis

    moderated by 5-HT (for review, Homberg and Lesch, 2011;

    Lesch, 2011). The molecular mechanisms by which environ-

    mental adversity impacts processing social cues and resulting

    emotional responses are not known, but are likely to include

    epigenetic programming of gene expression (Bartolomucci

    et al., 2010; Carola et al., 2008; van den Hove et al., 2011). In

    addition to its direct role in the regulation of gene transcription

    via receptor-mediated intracellular signaling-dependent activa-

    tion or inhibition of transcription factors, 5-HT also modulates

    synaptic plasticity by influencing epigenetic modification (DNA

    methylation, histone modifications) of genomic regions contain-

    ing transcription factors and through microRNA (miRNA)-facili-

    tated transcriptional repression via 5-HT receptor-mediated

    activation of intracellular signaling pathways converging on the

    transcriptional machinery of specific genes (e.g., Baudry et al.,

    2010; Figure 7). Epigenetic modification was therefore sug-

    gested as a potential mechanism for stabilizing gene expression

    that leads to persisting changes in the functional state of neurons

    required for long-term memory storage.

    miRNAs, a subclass of small RNA regulators that are involved

    in numerous cellular processes, including proliferation, differen-

    tiation, and plasticity (Krol et al., 2010; Millan, 2011), contribute

    to transcriptional and epigenetic regulation of gene expression

    during brain development and in differentiated neurons (Qureshi

    and Mehler, 2012; Saba and Schratt, 2010). Brain-specific miR-

    NAs constrain 5-HT-induced synaptic LTF through repression ofthe transcriptional activator CREB1 (Rajasethupathy et al.,

    2009). It has also recently been reported that another class of

    small noncoding regulatory RNAs, PIWI-interacting RNAs

    (piRNAs), are enriched in neurons of Aplysia and mouse and

    may have a role in spine morphogenesis (Lee et al., 2011; Raja-

    184 Neuron 76, October 4, 2012 2012 Elsevier Inc.sethupathy et al., 2012). Expression of several piRNAs is induced

    by 5-HT and PIWI/piRNA complexes moderate 5-HT-dependent

    methylation of CpG sites in the promoter of target genes, such as

    the plasticity-related transcriptional repressor CREB2. Together,

    these findings outline a small RNA-mediated gene regulatory

    mechanism for enhancing or constraining 5-HT-dependent

    LTF/LTP thus establishing enduring adjustments in mature

    neurons for the long-term encoding of memory and its cogni-

    tive-emotional reappraisal.

    Serotonin-Glutamate Interactionin Neurodevelopmental DisordersNeurodevelopmental disorders are generally characterized by

    severe impairments in the domains of attention, motivation,

    cognition, and emotion, display remarkable syndromal overlap,

    and persist across the life span. Multiple lines of evidence impli-

    cate serotonergic and glutamatergic pathway malfunction

    particularly in autism spectrum disorder (ASD), which is charac-

    terized by deficits in social cognition, communicative interaction,

    and emotional learning as well as by patterns of repetitive,

    restricted behaviors or interests and resistance to change (Du-

    rand et al., 2007; Grabrucker et al., 2011; Moessner et al., 2007).

    The role of 5-HT in ASD has been investigated with genetic,

    neuroimaging and biomarker approaches (for review, Pardo

    and Eberhart, 2007). Neuroimaging revealed that the peak in

    brain 5-HT synthesis capacity seen in typically developing

    infants at 2 years of age is absent in children with autism (Chan-

    dana et al., 2005). Reduction of 5-HT in dentatothalamocortical

    pathways, with simultaneous increases in the contralateral den-

    tate cerebellar nucleus as well as reduced 5-HT2A receptor

    binding in the cortical areas was reported (Murphy et al.,

    2006). These changes may reflect compromised formation of

    the 5-HT system with an increased number but dysmorphic

    manifestation of serotonin axons in terminal regions of the cortex

    (Azmitia et al., 2011). Moreover, elevated levels of 5-HT and

    decreased 5-HT2 receptor binding in the platelets have also

    been observed (Cook and Leventhal, 1996). Genetic studies

    have identified variants in 5-HT system-related genes, including

    5-HTT/SLC6A4 which also shows association with cortical gray

    matter volume and interaction with PTEN and neurotrophins,

    such as brain-derived neurotrophic factor (BDNF) (Page et al.,

    2009; Ren-Patterson et al., 2006; Ren-Patterson et al., 2005).

    Finally, pharmacological interventions with compounds acting

    on 5-HT2 receptors and SSRIs are effective in improving social

    cognition and interaction while decreasing aggressive and

    stereotyped behaviors in children with ASD (Cook and Leven-

    thal, 1996). Together, 5-HT system dysregulation coinciding

    with abnormalities in the glutamatergic pathway and their impact

    on brain development and plasticity supports a critical role of

    5-HT-glutamate interaction in the etiopathogenesis of autism

    and related disorders.

    Neurodevelopmental disorders display a complex genetic

    architecture where multiple common and rare genetic variants

    Reviewin interaction with environmental adversity contribute to risk.

    There is now replicated evidence that rare chromosomal dupli-

    cations and deletions known as copy-number variants (CNVs)

    are associated with ASD risk (for review, Abrahams and Gesch-

    wind, 2008; Devlin and Scherer, 2012) and that the chromosomal

  • an

    NeuronTr

    Transcriptional regulation

    Transcription factors

    Structural &

    functional proteins Epigenetic

    regulators

    piRNAs

    Reviewregions spanned by these CNVs show significant overlap

    with those implicated in attention-deficit/hyperactivity disorder

    (ADHD) and schizophrenia (Elia et al., 2012; Lesch et al., 2011;

    Lionel et al., 2011; Malhotra and Sebat, 2012; Talkowski et al.,

    2012; Williams et al., 2010b, 2012). Thus, it came as no surprise

    that these genome-wide analyses revealed risk genes encod-

    ing synaptic adhesion molecules (e.g., CDHs, NLGNs, NRXNs,

    and LPHNs), glutamate receptors (e.g., NMDARs, mGluRs) and

    their mediators of intracellular signaling pathways, as well as

    components of the PSD and activity-regulated cytoskeleton-

    associated protein complexes (e.g., SHANKs). In ASD, CNV

    screening and deep sequencing are rapidly identifying genes

    for further characterization. These approaches have implicated,

    among others, CDH810, CDH13, NLGN3, NLGN4, SHANK13,

    NRXN1,NRXN3, ASTN2,DPP6, andCNTNAP2 as affecting ASD

    risk (Devlin and Scherer, 2012; Pagnamenta et al., 2011; Sanders

    et al., 2011; Singh et al., 2010; Wang et al., 2009). Some rare,

    highly penetrant mutations appear to be monogenic causes of

    ASD. Moreover, large-scale whole-exome sequencing is

    currently identifying numerous rare single nucleotide variants

    mRNA

    Epigenetic pattern DNA Methylation

    Me

    Ac Histone modifications

    Figure 7. Regulatory Circuits of Gene ExpressionEpigenetic mechanisms including DNA methylation and histone modifications afraction of small noncoding regulatory RNAs, regulate gene expression patternepigenetic modifications and miRNAs control each other to form regulatory circregulation of the transcriptional machinery via receptor-mediated intracellular smodulates synaptic plasticity by influencing epigenetic patterns of genomic regrepression via 5-HT receptor-mediated activation of intracellular signaling pathwscription

    +

    - Posttranscriptional

    repression

    Epigenetically regulated microRNAs (SNVs) potentially be associated with de novo and inherited

    ASD (Neale et al., 2012; ORoak et al., 2012; Sanders et al.,

    2012).

    Since individuals with severe disorders particularly of the

    autism or schizophrenia spectrum may have developed as ex-

    pected but at a certain stage present with indicators of regres-

    sion in cognitive, emotional, and psychosocial capacities,

    neurodevelopmental disorders may not arise from a deleterious

    impact on initial synapse formation but rather on synaptic re-

    modeling in the course of the brains maturation and subsequent

    consolidation processes of circuit connectivity. The core symp-

    toms of neurodevelopmental disorders likely arise from a defi-

    ciency in the multifaceted crosstalk among numerous synaptic

    adhesion molecules, both at the extracellular level and at the

    level of their intracellular signaling pathways.

    Based on the contribution of adhesion molecules to synaptic

    remodeling and circuit maturation in neurodevelopmental dis-

    orders, the contribution of NRXNs and NLGNs to cognitive

    function and synaptic plasticity was also studied in genetically

    modified mouse models. Mice constitutively deficient for Nlgn1

    mRNA cleavage

    Translational repression

    Deadenylation microRNAs

    s well as microRNAs (miRNAs) and PIWI-interacting RNAs (piRNAs), anothers at both the transcriptional and posttranscriptional level. At the same time,uits and to maintain physiological functions. In addition of its direct role in theignaling-dependent activation or inhibition of transcription factors, 5-HT alsoions containing transcription factors or by miRNAs-facilitated transcriptionalays converging on the transcriptional machinery of specific genes.

    Neuron 76, October 4, 2012 2012 Elsevier Inc. 185

  • Neuronrevealed that NGLNs are essential for lateral trafficking of

    NMDA receptors to postsynaptic site and maintaining NMDA

    receptor-mediated currents, whereas a humanized mouse

    model with a knockin of a NLGN3 mutation was reported to

    display autism-related behavioral abnormalities (Tabuchi et al.,

    2007). In contrast, Nrxn-1a knockout mice exhibit enhanced

    motor learning capacities, despite deficient glutamatergic

    transmission (Etherton et al., 2009). Together, Nrxn and Nlgn

    inactivation fails to change synapse number, suggesting that

    both moderate synaptic remodeling and maturation rather

    than initial synapse formation. In support of a contribution of

    adhesion molecules to the activity-dependent modification of

    developing neural circuits, in vitro approaches revealed that

    inhibition of NMDA receptors suppresses the synaptogenic

    activity of NLGN1 (Chubykin et al., 2007). Mutations in SHANK3

    (Durand et al., 2007; Grabrucker et al., 2011; Moessner et al.,

    2007) are thought to result in modifications of dendritic spine

    morphology via an actin-dependent mechanism (Durand et al.,

    2012), likely to result in defects at striatal synapses and cortico-

    striatal circuits that were reported in Shank3 mutant mice (Peca

    et al., 2011).

    Linking 5-HT Receptor-mGluR Crosstalk to Defectsin Synaptic PlasticityTranssynaptic signaling mediated by mGluR5 modulates effi-

    ciency and timing of excitatory transmission in a behaviorally

    relevant manner. Group I, II, and III mGluRmembers are required

    for different modes of pre- and postsynaptic short- and long-

    term plasticity. Given the target-specific distribution of mGluRs,

    such that synaptic input from one presynaptic neuron is modu-

    lated by different receptors at each of its postsynaptic targets,

    mGluRs provide a mechanism for synaptic specialization of glu-

    tamatergic transmission. Interactions between 5-HT receptors

    and mGluRs have also been identified. For example, mGluR2

    interacts through specific transmembrane helix domains with

    the 5-HT1A receptor to form functional complexes in cortex,

    thus triggering cellular responses in disorders of cognitive pro-

    cessing and in response to pharmacological intervention

    (Gonzalez-Maeso et al., 2008).

    Although mGluR5 was previously implicated in neurodevelop-

    mental disorders (Auerbach et al., 2011; Devon et al., 2001;

    Schumann et al., 2008; Wang et al., 2010), a recent genome-

    wide screen showed that the GRM gene family encoding

    mGluRs, most frequently GRM5, and genes interacting with it

    are enriched for CNVs in ADHD (Elia et al., 2012; Lesch et al.,

    2012b). ADHD is characterized by developmentally inappro-

    priate inattention, hyperactivity, increased impulsivity and emo-

    tional dysregulation with a specific constellation of deficits in

    motivation, working memory and cognitive control of executive

    functions, thus displaying syndromal overlap with ASD. Other

    CNV findings concerned GRM1 duplications, GRM7 deletions,

    and GRM8 deletions. Overall the findings indicate that up to

    10% of individuals with ADHD may be enriched for mGluRnetwork variants. Several of these genes play a central role in

    the process of neurogenesis, synaptic transmission and network

    connectivity that has been argued to be defective in ADHD.

    Specifically, mGluRs modulate mRNA generation, alternative

    splicing and translation, processes known to influence

    186 Neuron 76, October 4, 2012 2012 Elsevier Inc.circuitry-specific formation, activity and plasticity of synapses

    (Bockaert et al., 2010; Knafo and Esteban, 2012).

    Disruption of frontostriatal circuitries which are involved in

    motor control and action learning, is thought to represent a

    specific characteristic of ADHD pathophysiology (Cubillo et al.,

    2012; de Zeeuw et al., 2012). Enhanced short-range connectivity

    within motivation-reward networks and their decreased connec-

    tivity with structures comprising the default-mode and dorsal

    attention networks have been reported, indicating impaired

    crosstalk among cognitive control and reward pathways that

    may reflect attentional andmotivational deficits in ADHD (Tomasi

    and Volkow, 2012; Volkow et al., 2012). Since it is abundantly ex-

    pressed in dendritic spines of structural units of the frontostriatal

    circuit including nucleus accumbens, dorsal striatum and PFC,

    mGluR5 not only interacts with signaling of dopamine and

    5-HT receptors but also with NMDA receptors, resulting in recip-

    rocal and agonist-independent inhibition of the two receptors

    (Perroy et al., 2008). While mGluR5 is confined to the periphery

    of the synapse, NMDA receptors are located vis-a`-vis of the

    glutamate release site in the PSD comprising the multiprotein

    HOMER-SHANK-GKAP-PSD-95 scaffolding complex physically

    and functionally linking the two receptors (Fagni et al., 2008).

    Moreover, the nucleus accumbens and dorsal striatum receive

    extensive serotonergic input mediated by a multitude of 5-HT

    receptors including subtypes 5-HT1-4 (Figure 2). 5-HT activates

    5-HT1B receptors resulting in a cAMP-dependent LTD-associ-

    ated decrease of glutamate release and striatal output (Mathur

    et al., 2011; Navailles and De Deurwaerdere, 2011). This 5-HT-

    induced LTD is independent of dopamine, suggesting that sero-

    tonergic and dopaminergic signaling pathways both interact in

    corticostriatal circuit plasticity. In line with these presumed

    molecular mechanisms, both pharmacological inhibition of

    mGlurR5 or targeted inactivation of its gene Grm5 result in loco-

    motor hyperactivity and reduced habituation to novelty (Halber-

    stadt et al., 2011; Kachroo et al., 2005). Deficits in spatial learning

    as well as acquisition and retrieval of stimulus-outcome memo-

    ries in a fear conditioning paradigm have also been reported

    (Jia et al., 2001; Xu et al., 2009). Electrophysiological studies in

    Grm5 knockout mice revealed sensorimotor gating deficits sug-

    gesting a key role for this gene in the modulation of hippocampal

    NMDA receptor-dependent synaptic plasticity (Jia et al., 1998).

    Dissection and characterization of the molecular components

    of these transsynaptic signaling interfaces and their involvement

    in the modulatory action of 5-HT on synaptic plasticity is likely to

    give better insight into the pathogenesis of neurodevelopmental

    disorders and to provide novel targets for translation into inter-

    ventional strategies.

    Conclusion and OutlookOur understanding of how 5-HT-dependent modulation of circuit

    configuration influences social cognition and emotional learning

    has been enhanced by recent insight into the molecular

    machinery that connects pre- and postsynaptic neurons and

    Reviewthe cellular mechanisms of synapse formation and plasticity.

    However, we have made only the first few steps on the long

    and winding road toward an understanding of the neural mech-

    anisms underlying cognition-emotion continuum as the funda-

    mental basis of effective social functioning (Pessoa, 2008), and

  • toric population expansion andmigration, agricultural revolution,

    Neuronindustrialization, and urbanization of life styles (Lupski et al.,

    2011; McClellan and King, 2010), which complements the

    evidence for a link between neural plasticity and the multidimen-

    sional cognitive and emotional processes of decision-making

    (Canli and Lesch, 2007; Frith and Singer, 2008; Steinbeis et al.,

    2012). Although the fusion of humanities, social sciences and

    neurosciences is under way, the transition from complex corre-

    lations and interactions to applicable prediction is the genuine

    challenge.

    ACKNOWLEDGMENTS

    We apologize to colleagues whose work could not be cited due to spacelimitations. The writing of this article and the authors related research weresupported by the Deutsche Forschungsgemeinschaft (SFB 581/B9, SFBTRR 58/A1 and A5, KFO 125). The authors thank J. Stilla and G. Lesch forassistance in generating graphical material. The authors are also grateful toC. Gross for his critical comments.

    REFERENCES

    Abrahams, B.S., andGeschwind, D.H. (2008). Advances in autism genetics: onthe threshold of a new neurobiology. Nat. Rev. Genet. 9, 341355.

    Albert, P.R. (2012). Transcriptional regulation of the 5-HT1A receptor:implications for mental illness. Philos. Trans. R. Soc. Lond. B Biol. Sci. 367,24022415.

    Amodio, D.M., and Frith, C.D. (2006). Meeting of minds: the medial frontalcortex and social cognition. Nat. Rev. Neurosci. 7, 268277.

    Ansorge, M.S., Zhou, M., Lira, A., Hen, R., and Gingrich, J.A. (2004). Early-lifeblockade of the 5-HT transporter alters emotional behavior in adult mice.Science 306, 879881.

    Auerbach, B.D., Osterweil, E.K., and Bear, M.F. (2011). Mutations causingsyndromic autism define an axis of synaptic pathophysiology. Nature 480,the contribution of 5-HT signaling to these mechanisms. Yet, the

    potential impact of 5-HT-modulated synaptic plasticity on social

    cognition and emotionality is currently transcending the bound-

    aries of behavioral genetics, molecular neurobiology and cogni-

    tive neuroscience to embrace biosocial science, thus creating

    the framework for a biosocial brain (Lesch, 2007). Detailed

    analyses of human genomes, together with a wide range of other

    species, has revealed an unexpected magnitude of variation in

    individuals, reflecting remarkable genomic plasticity (Gerstein

    et al., 2012; Keinan and Clark, 2012; Wolf and Linden, 2012).

    These genetic analyses are contributing fundamentally to the

    knowledge of how humans have evolved, how we (mal)function,

    and why we suffer from or resist to disease. Genetic approaches

    have matured to explore the underestimated wealth of genetic

    variation among humans and its influence on interindividual

    differences and the relative impact of neural and environmental

    determinants on cognition, emotionality, and behavior. The

    science of the biosocial brain increasingly uses neuroimaging

    to study the neural basis of complex behavior, examining such

    phenomena as social conformity, empathy, trust and altruism

    (Carr et al., 2003) as well as evolutionary (epi)genetics of prehis-

    Review6368.

    Azmitia, E.C., and Whitaker-Azmitia, P.M. (1997). Development and adultplasticity of serotonergic neurons and their target cells. In Serotonergicneurons and 5-HT receptors in the CNS, H.G. Baumgarten and M. Gothert,eds. (Berlin, Heidelberg, New York: Springer), pp. 139.Azmitia, E.C., Singh, J.S., and Whitaker-Azmitia, P.M. (2011). Increasedserotonin axons (immunoreactive to 5-HT transporter) in postmortem brainsfrom young autism donors. Neuropharmacology 60, 13471354.

    Barnes, N.M., and Sharp, T. (1999). A review of central 5-HT receptors andtheir function. Neuropharmacology 38, 10831152.

    Barr, C.S., Newman, T.K., Shannon, C., Parker, C., Dvoskin, R.L., Becker,M.L., Schwandt, M., Champoux, M., Lesch, K.P., Goldman, D., et al. (2004).Rearing condition and rh5-HTTLPR interact to influence limbic-hypotha-lamic-pituitary-adrenal axis response to stress in infant macaques. Biol.Psychiatry 55, 733738.

    Bartolomucci, A., Carola, V., Pascucci, T., Puglisi-Allegra, S., Cabib, S., Lesch,K.P., Parmigiani, S., Palanza, P., and Gross, C. (2010). Increased vulnerabilityto psychosocial stress in heterozygous serotonin transporter knockout mice.Dis. Model. Mech. 3, 459470.

    Baudry, A., Mouillet-Richard, S., Schneider, B., Launay, J.M., and Kellermann,O. (2010). miR-16 targets the serotonin transporter: a new facet for adaptiveresponses to antidepressants. Science 329, 15371541.

    Becamel, C., Gavarini, S., Chanrion, B., Alonso, G., Galeotti, N., Dumuis, A.,Bockaert, J., and Marin, P. (2004). The serotonin 5-HT2A and 5-HT2C recep-tors interact with specific sets of PDZ proteins. J. Biol. Chem. 279, 2025720266.

    Bennett, A.J., Lesch, K.P., Heils, A., Long, J.C., Lorenz, J.G., Shoaf, S.E.,Champoux, M., Suomi, S.J., Linnoila, M.V., and Higley, J.D. (2002). Earlyexperience and serotonin transporter gene variation interact to influenceprimate CNS function. Mol. Psychiatry 7, 118122.

    Bennett-Clarke, C.A., Chiaia, N.L., and Rhoades, R.W. (1996). Thalamocorticalafferents in rat transiently express high-affinity serotonin uptake sites. BrainRes. 733, 301306.

    Benson, D.L., Schnapp, L.M., Shapiro, L., and Huntley, G.W. (2000). Makingmemories stick: cell-adhesion molecules in synaptic plasticity. Trends CellBiol. 10, 473482.

    Bethea, C.L., and Reddy, A.P. (2012a). Effect of ovarian steroids on geneexpression related to synapse assembly in serotonin neurons of macaques.J. Neurosci. Res. 90, 13241334.

    Bethea, C.L., and Reddy, A.P. (2012b). Ovarian steroids increase glutamater-gic related gene expression in serotonin neurons of macaques. Mol. Cell. Neu-rosci. 49, 251262.

    Blakemore, S.J. (2008). The social brain in adolescence. Nat. Rev. Neurosci. 9,267277.

    Bockaert, J., Perroy, J., Becamel, C., Marin, P., and Fagni, L. (2010). GPCR in-teracting proteins (GIPs) in the nervous system: Roles in physiology andpathologies. Annu. Rev. Pharmacol. Toxicol. 50, 89109.

    Boucard, A.A., Ko, J., and Sudhof, T.C. (2012). High affinity neurexin binding tocell adhesion G-protein-coupled receptor CIRL1/latrophilin-1 produces anintercellular adhesion complex. J. Biol. Chem. 287, 93999413.

    Bredt, D.S., and Nicoll, R.A. (2003). AMPA receptor trafficking at excitatorysynapses. Neuron 40, 361379.

    Calizo, L.H., Akanwa, A., Ma, X., Pan, Y.Z., Lemos, J.C., Craige, C., Heemstra,L.A., and Beck, S.G. (2011). Raphe serotonin neurons are not homogenous:electrophysiological, morphological and neurochemical evidence. Neurophar-macology 61, 524543.

    Canli, T., and Lesch, K.P. (2007). Long story short: the serotonin transporter inemotion regulation and social cognition. Nat. Neurosci. 10, 11031109.

    Canli, T., Qiu, M., Omura, K., Congdon, E., Haas, B.W., Amin, Z., Herrmann,M.J., Constable, R.T., and Lesch, K.P. (2006). Neural correlates of epigenesis.

    Proc. Natl. Acad. Sci. USA 103, 1603316038.

    Carola, V., Frazzetto, G., Pascucci, T., Audero, E., Puglisi-Allegra, S., Cabib,S., Lesch, K.P., and Gross, C. (2008). Identifying molecular substrates ina mouse model of the serotonin transporter x environment risk factor foranxiety and depression. Biol. Psychiatry 63, 840846.

    Neuron 76, October 4, 2012 2012 Elsevier Inc. 187

  • NeuronCarr, L., Iacoboni, M., Dubeau, M.C., Mazziotta, J.C., and Lenzi, G.L. (2003).Neural mechanisms of empathy in humans: a relay from neural systems forimitation to limbic areas. Proc. Natl. Acad. Sci. USA 100, 54975502.

    Chandana, S.R., Behen, M.E., Juhasz, C., Muzik, O., Rothermel, R.D.,Mangner, T.J., Chakraborty, P.K., Chugani, H.T., and Chugani, D.C. (2005).Significance of abnormalities in developmental trajectory and asymmetry ofcortical serotonin synthesis in autism. Int. J. Dev. Neurosci. 23, 171182.

    Chanrion, B., Mannoury la Cour, C., Bertaso, F., Lerner-Natoli, M., Freissmuth,M., Millan, M.J., Bockaert, J., and Marin, P. (2007). Physical interactionbetween the serotonin transporter and neuronal nitric oxide synthase underliesreciprocal modulation of their activity. Proc. Natl. Acad. Sci. USA 104, 81198124.

    Choi, Y.B., Li, H.L., Kassabov, S.R., Jin, I., Puthanveettil, S.V., Karl, K.A., Lu,Y., Kim, J.H., Bailey, C.H., and Kandel, E.R. (2011). Neurexin-neuroligintranssynaptic interaction mediates learning-related synaptic remodeling andlong-term facilitation in aplysia. Neuron 70, 468481.

    Chubykin, A.A., Atasoy, D., Etherton, M.R., Brose, N., Kavalali, E.T., Gibson,J.R., and Sudhof, T.C. (2007). Activity-dependent validation of excitatoryversus inhibitory synapses by neuroligin-1 versus neuroligin-2. Neuron 54,919931.

    Cook, E.H., and Leventhal, B.L. (1996). The serotonin system in autism. Curr.Opin. Pediatr. 8, 348354.

    Cordes, S.P. (2005). Molecular genetics of the early development of hindbrainserotonergic neurons. Clin. Genet. 68, 487494.

    Cruz, D.A., Eggan, S.M., Azmitia, E.C., and Lewis, D.A. (2004). Serotonin1Areceptors at the axon initial segment of prefrontal pyramidal neurons inschizophrenia. Am. J. Psychiatry 161, 739742.

    Cubillo, A., Halari, R., Smith, A., Taylor, E., and Rubia, K. (2012). A review offronto-striatal and fronto-cortical brain abnormalities in children and adultswith attention deficit hyperactivity disorder (ADHD) and new evidence fordysfunction in adults with ADHD during motivation and attention. Cortex 48,194215.

    Dahlstrom, A., and Fuxe, K. (1964). Localization of monoamines in the lowerbrain stem. Experientia 20, 398399.

    Dalva, M.B., McClelland, A.C., and Kayser, M.S. (2007). Cell adhesion mole-cules: signalling functions at the synapse. Nat. Rev. Neurosci. 8, 206220.

    Daubert, E.A., and Condron, B.G. (2010). Serotonin: a regulator of neuronalmorphology and circuitry. Trends Neurosci. 33, 424434.

    de Almeida, J., and Mengod, G. (2007). Quantitative analysis of glutamatergicand GABAergic neurons expressing 5-HT(2A) receptors in human and monkeyprefrontal cortex. J. Neurochem. 103, 475486.

    de Almeida, J., and Mengod, G. (2008). Serotonin 1A receptors in human andmonkey prefrontal cortex are mainly expressed in pyramidal neurons and in aGABAergic interneuron subpopulation: implications for schizophrenia and itstreatment. J. Neurochem. 107, 488496.

    De Blasi, A., Conn, P.J., Pin, J., and Nicoletti, F. (2001). Molecular determi-nants of metabotropic glutamate receptor signaling. Trends Pharmacol. Sci.22, 114120.

    de Wit, J., Sylwestrak, E., OSullivan, M.L., Otto, S., Tiglio, K., Savas, J.N.,Yates, J.R., 3rd, Comoletti, D., Taylor, P., and Ghosh, A. (2009). LRRTM2interacts with Neurexin1 and regulates excitatory synapse formation. Neuron64, 799806.

    de Zeeuw, P., Mandl, R.C., Hulshoff Pol, H.E., van Engeland, H., and Durston,S. (2012). Decreased frontostriatal microstructural organization in attentiondeficit/hyperactivity disorder. Hum. Brain Mapp. 33, 19411951.

    Derkach, V.A., Oh, M.C., Guire, E.S., and Soderling, T.R. (2007). Regulatorymechanisms of AMPA receptors in synaptic plasticity. Nat. Rev. Neurosci. 8,

    101113.

    Devlin, B., and Scherer, S.W. (2012). Genetic architecture in autism spectrumdisorder. Curr. Opin. Genet. Dev. 22, 229237.

    Devon, R.S., Anderson, S., Teague, P.W., Muir, W.J., Murray, V., Pelosi, A.J.,Blackwood, D.H., and Porteous, D.J. (2001). The genomic organisation of the

    188 Neuron 76, October 4, 2012 2012 Elsevier Inc.metabotropic glutamate receptor subtype 5 gene, and its association withschizophrenia. Mol. Psychiatry 6, 311314.

    Durand, C.M., Betancur, C., Boeckers, T.M., Bockmann, J., Chaste, P.,Fauchereau, F., Nygren, G., Rastam, M., Gillberg, I.C., Anckarsater, H., et al.(2007). Mutations in the gene encoding the synaptic scaffolding proteinSHANK3 are associated with autism spectrum disorders. Nat. Genet. 39,2527.

    Durand, C.M., Perroy, J., Loll, F., Perrais, D., Fagni, L., Bourgeron, T.,Montcouquiol, M., and Sans, N. (2012). SHANK3mutations identified in autismlead to modification of dendritic spine morphology via an actin-dependentmechanism. Mol. Psychiatry 17, 7184.

    Elia, J., Glessner, J.T., Wang, K., Takahashi, N., Shtir, C.J., Hadley, D.,Sleiman, P.M.A., Zhang, H., Kim, C.E., Robison, R., et al. (2012). Genome-wide copy number variation study associates metabotropic glutamatereceptor gene networks with attention deficit hyperactivity disorder. Nat.Genet. 44, 7884.

    Erzurumlu, R.S., and Gaspar, P. (2012). Development and critical periodplasticity of the barrel cortex. Eur. J. Neurosci. 35, 15401553.

    Etherton, M.R., Blaiss, C.A., Powell, C.M., and Sudhof, T.C. (2009). Mouseneurexin-1alpha deletion causes correlated electrophysiological and behav-ioral changes consistent with cognitive impairments. Proc. Natl. Acad. Sci.USA 106, 1799818003.

    Fagni, L., Bertaso, F., Perroy, J., and Ango, F. (2008). Unexpected roles ofscaffolding proteins in receptor patho-physiological functions. J. Integr.Neurosci. 7, 211224.

    Frith, C.D., and Frith, U. (2012). Mechanisms of social cognition. Annu. Rev.Psychol. 63, 287313.

    Frith, C.D., and Singer, T. (2008). The role of social cognition in decisionmaking. Philos. Trans. R. Soc. Lond. B Biol. Sci. 363, 38753886.

    Fu, W., Le Maitre, E., Fabre, V., Bernard, J.F., David Xu, Z.Q., and Hokfelt, T.(2010). Chemical neuroanatomy of the dorsal raphe nucleus and adjacentstructures of the mouse brain. J. Comp. Neurol. 518, 34643494.

    Gaspar, P., Cases, O., and Maroteaux, L. (2003). The developmental role ofserotonin: news from mouse molecular genetics. Nat. Rev. Neurosci. 4,10021012.

    Gerstein, M.B., Kundaje, A., Hariharan, M., Landt, S.G., Yan, K.K., Cheng, C.,Mu, X.J., Khurana, E., Rozowsky, J., Alexander, R., et al. (2012). Architectureof the human regulatory network derived from ENCODE data. Nature 489,91100.

    Gonzalez-Maeso, J., Ang, R.L., Yuen, T., Chan, P., Weisstaub, N.V., Lopez-Gimenez, J.F., Zhou, M., Okawa, Y., Callado, L.F., Milligan, G., et al. (2008).Identification of a serotonin/glutamate receptor complex implicated inpsychosis. Nature 452, 9397.

    Grabrucker, A.M., Schmeisser, M.J., Schoen, M., and Boeckers, T.M. (2011).Postsynaptic ProSAP/Shank scaffolds in the cross-hair of synaptopathies.Trends Cell Biol. 21, 594603.

    Gross, C., and Hen, R. (2004). The developmental origins of anxiety. Nat. Rev.Neurosci. 5, 545552.

    Gross, C., Zhuang, X., Stark, K., Ramboz, S., Oosting, R., Kirby, L., Santarelli,L., Beck, S., and Hen, R. (2002). Serotonin1A receptor acts during develop-ment to establish normal anxiety-like behaviour in the adult. Nature 416,396400.

    Gutknecht, L., Kriegebaum, C., Waider, J., Schmitt, A., and Lesch, K.P. (2009).Spatio-temporal expression of tryptophan hydroxylase isoforms in murine andhuman brain: convergent data from Tph2 knockout mice. Eur. Neuropsycho-pharmacol. 19, 266282.

    Gutknecht, L., Araragi, N., Merker, S., Waider, J., Sommerlandt, F.M., Mlinar,

    ReviewB., Baccini, G., Mayer, U., Proft, F., Hamon, M., et al. (2012). Impacts of brainserotonin deficiency following Tph2 inactivation on development and rapheneuron serotonergic specification. PLoS ONE 7, e43157.

    Halberstadt, A.L., Lehmann-Masten, V.D., Geyer, M.A., and Powell, S.B.(2011). Interactive effects of mGlu5 and 5-HT2A receptors on locomotoractivity in mice. Psychopharmacology (Berl.) 215, 8192.

  • NeuronHay-Schmidt, A. (2000). The evolution of the serotonergic nervous system.Proc. Biol. Sci. 267, 10711079.

    Hendricks, T., Francis, N., Fyodorov, D., and Deneris, E.S. (1999). The ETSdomain factor Pet-1 is an early and precisemarker of central serotonin neuronsand interacts with a conserved element in serotonergic genes. J. Neurosci. 19,1034810356.

    Hensler, J.G. (2006). Serotonergic modulation of the limbic system. Neurosci.Biobehav. Rev. 30, 203214.

    Holtmaat, A., and Svoboda, K. (2009). Experience-dependent structuralsynaptic plasticity in the mammalian brain. Nat. Rev. Neurosci. 10, 647658.

    Homberg, J.R., and Lesch, K.P. (2011). Looking on the bright side of serotonintransporter gene variation. Biol. Psychiatry 69, 513519.

    Hornung, J.P., Fritschy, J.M., and Tork, I. (1990). Distribution of two morpho-logically distinct subsets of serotoninergic axons in the cerebral cortex of themarmoset. J. Comp. Neurol. 297, 165181.

    Inaba, M., Maruyama, T., Yoshimura, Y., Hosoi, H., and Komatsu, Y. (2009).Facilitation of low-frequency stimulation-induced long-term potentiation byendogenous noradrenaline and serotonin in developing rat visual cortex.Neurosci. Res. 64, 191198.

    Jacobsen, K.X., Czesak, M., Deria, M., Le Francois, B., and Albert, P.R. (2011).Region-specific regulation of 5-HT1A receptor expression by Pet-1-depen-dent mechanisms in vivo. J. Neurochem


Recommended